EP4122914A1 - Compound and method for preparation of lisdexamfetamine - Google Patents

Compound and method for preparation of lisdexamfetamine Download PDF

Info

Publication number
EP4122914A1
EP4122914A1 EP21187139.7A EP21187139A EP4122914A1 EP 4122914 A1 EP4122914 A1 EP 4122914A1 EP 21187139 A EP21187139 A EP 21187139A EP 4122914 A1 EP4122914 A1 EP 4122914A1
Authority
EP
European Patent Office
Prior art keywords
formula
group
salt
compound represented
acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP21187139.7A
Other languages
German (de)
French (fr)
Other versions
EP4122914B1 (en
Inventor
Chih-Wei Tseng
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
SCI Pharmtech Inc
Original Assignee
SCI Pharmtech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by SCI Pharmtech Inc filed Critical SCI Pharmtech Inc
Priority to EP21187139.7A priority Critical patent/EP4122914B1/en
Priority to EP23156189.5A priority patent/EP4215517A1/en
Publication of EP4122914A1 publication Critical patent/EP4122914A1/en
Application granted granted Critical
Publication of EP4122914B1 publication Critical patent/EP4122914B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/08Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms
    • C07C271/10Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C271/22Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms to carbon atoms of hydrocarbon radicals substituted by carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C231/00Preparation of carboxylic acid amides
    • C07C231/12Preparation of carboxylic acid amides by reactions not involving the formation of carboxamide groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C269/00Preparation of derivatives of carbamic acid, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C269/06Preparation of derivatives of carbamic acid, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups by reactions not involving the formation of carbamate groups
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • the present disclosure relates to compounds for preparing lisdexamfetamine, and to methods for preparing lisdexamfetamine from the compounds.
  • Lisdexamfetamine dimesylate i.e., (2S)-2,6-diamino- N -[(1S)-1-methyl-2-phenylethyl] hexanamide dimethanesulfonate under the brand name of Vyvanse
  • ADHD attention deficit hyperactivity disorder
  • BED moderate to severe binge eating disorder
  • U.S. Patent Publication No. 2012/0157706 discloses a method for preparation of lisdexamfetamine dimesylate from D-amphetamine. The method involves reacting D-amphetamine with (S)-2,5-dioxopyrrolidin-1-yl 2,6-bis(benzyloxycarbonylamino) hexanoate to form a lysine amphetamine bearing benzylcarbamate protecting groups.
  • the lysine amphetamine bearing the benzylcarbamate protecting groups is then converted to lisdexamfetamine dimesylate by catalytic hydrogenation to remove the benzylcarbamate protecting groups, followed by subsequent addition of methanesulfonic acid to generate the lisdexamfetamine dimesylate.
  • International Patent Publication No. WO 2010/042120 discloses a process for preparation of lisdexamfetamine by reacting D-amphetamine with (S)-2,6-bis((tert-butoxycarbonyl)amino)hexanoic acid in the presence of an alkylphosphonic acid anhydride (e.g., T3P) as a coupling agent under a base condition including a solvent, followed by adding methanesulfonic acid to remove the tert-butoxycarbonyl protecting groups and thus generate lisdexamfetamine dimesylate.
  • an alkylphosphonic acid anhydride e.g., T3P
  • methanesulfonic acid to remove the tert-butoxycarbonyl protecting groups and thus generate lisdexamfetamine dimesylate.
  • WO 2013/011526 discloses another process for preparation of lisdexamfetamine by reacting D-amphetamine with (S)-2,6-bis((tert-butoxycarbonyl)amino)hexanoic acid in the presence of isobutyl chloroformate as a coupling agent under a base condition including a solvent.
  • WO 2010/148305 discloses a process for preparation of lisdexamfetamine starting from L-norephedrine.
  • L-norephedrine is converted to the corresponding chloro-D-amphetamine hydrochloride by thionyl chloride, and the resulting chloro-D-amphetamine hydrochloride is coupled with (S)-2,6-bis(2,2,2-trifluoroacetamido)hexanoic acid to obtain an N,N'-bis-trifluoroacetyl-protected chloro-lisdexamfetamine intermediate.
  • N,N'-bis-trifluoroacetyl-protected chloro-lisdexamfetamine intermediate is converted to N,N'-bis-trifluoroacetyl-protected lisdexamfetamine by removal of chlorine with catalytic hydrogenation.
  • lisdexamfetamine dimesylate is obtained by subjecting N,N'-bis-trifluoroacetyl-protected lisdexamfetamine to a hydrolysis reaction by methanesulfonic acid.
  • U.S. Patent Publication No. 2016/0376618 discloses a method for preparation of lisdexamfetamine from amfetamine, comprising reacting amfetamine with N,N'-bis-acyllysine ester and an enzyme catalyst to form a lisdexamfetamine stereoisomer, followed by converting the stereoisomer to lisdexamfetamine by treating with tetrakis(triphenylphosphine)palladium to remove an allyloxycarbonyl protecting group. Subsequently, by addition of methanesulfonic acid, lisdexamfetamine dimesylate is obtained.
  • D-amphetamine also known as dextroamphetamine
  • dextroamphetamine is a Schedule II drug defined by the United States Controlled Substances Act. Therefore, special licenses, handling procedures, and compliance with governmental regulatory provisions are required for using large quantities of the controlled substance.
  • chloro-D-amphetamine or phenylaziridine is used as a starting material to replace the D-amphetamine. However, these processes involve additional steps of inserting a chloro group and thereafter removing the chloro group. In addition, multi-steps are required for synthesis of phenylaziridine.
  • the present disclosure provides a method for preparing lisdexamfetamine represented by Formula (I) below and a salt thereof:
  • the present disclosure provides a compound represented by Formula (VI) below or a salt thereof for preparing lisdexamphetamine or a salt thereof: wherein R 1 is hydrogen, an aliphatic group, C 1 -C 6 alkoxyl, a C 1 -C 6 alkylamino group, or an aromatic group, and PG is an amine protecting group.
  • R 1 is hydrogen, an aliphatic group, C 1 -C 6 alkoxyl, a C 1 -C 6 alkylamino group, or an aromatic group
  • PG is an amine protecting group.
  • R 1 is methyl.
  • PG is benzyloxycarbonyl.
  • the compound for preparing lisdexamphetamine or a salt thereof is represented by Formula (VIa) below: wherein Cbz refers to benzyloxycarbonyl.
  • the method for preparing lisdexamfetamine or a salt thereof provided herein comprises reducing an O-acylation group of an intermediate compound represented by Formula (VI) or a salt thereof through a catalytic hydrogenation to obtain the lisdexamphetamine or the salt thereof: wherein R 1 is hydrogen, an aliphatic group, C1-C6 alkoxyl, a C1-C6 alkylamino group, or an aromatic group, and PG is an amine protecting group.
  • R 1 is hydrogen, an aliphatic group, C1-C6 alkoxyl, a C1-C6 alkylamino group, or an aromatic group
  • PG is an amine protecting group.
  • the method further comprises reacting a compound represented by Formula (V) or a salt thereof with an acylation reagent to obtain the intermediate compound represented by Formula (VI): wherein R 1 and PG are defined as above.
  • the method further comprises coupling L-norephedrine represented by Formula (II) or a salt thereof and a diamino protected L-lysine represented by Formula (IV) or a salt thereof to obtain the compound represented by Formula (V): wherein PG is defined as above.
  • the coupling L-norephedrine or the salt thereof and the diamino protected L-lysine or the salt thereof is carried out by reaction with an acid activating reagent: wherein PG is defined as above.
  • the method further comprises reacting L-lysine represented by Formula (III) below with an amine protecting group in the presence of a protection reagent and a solvent to obtain the diamino protected L-lysine represented by Formula (IV): wherein PG is the amine protecting group.
  • the method further comprises contacting the lisdexamphetamine with methanesulfonic acid to obtain lisdexamfetamine dimesylate.
  • compositions, methods, and respective component(s) thereof are included in the present disclosure, yet open to the inclusion of unspecified elements.
  • a composition, mixture, process or method that comprises a list of elements or actions is not necessarily limited to only those elements or actions, but may include other elements or actions not expressly listed, or inherent to such composition, mixture, process, or method.
  • first As used herein, the terms “first,” “second,” “third,” and the like (if present) are used to distinguish similar objects and are not necessarily used to describe a particular order. It should be understood that the objects are interchangeable at an appropriate time.
  • the term “about” generally means within 10%, 5%, 1%, or 0.5% of a given value or range. Alternatively, the term “about” means within an acceptable standard error of the mean when considered by one of ordinary skill in the art. Unless otherwise expressly specified, all of the numerical ranges, amounts, values and percentages such as those for quantities of materials, durations of time periods, temperatures, operating conditions, ratios of amounts, and the likes disclosed herein should be understood as modified in all instances by the term "about.”
  • the present disclosure is directed to an intermediate compound and a method for preparing lisdexamphetamine or a salt thereof.
  • the intermediate compound for the preparation method is represented by Formula (VI) below: or a salt thereof, wherein R 1 is hydrogen, an aliphatic group, C1-C6 alkoxyl, a C1-C6 alkylamino group, or an aromatic group, and PG is an amine protecting group.
  • R 1 is hydrogen, an aliphatic group, C1-C6 alkoxyl, a C1-C6 alkylamino, or an aromatic group
  • PG is an amine protecting group
  • the method for preparing lisdexamfetamine represented by Formula (I) and a salt thereof may start from preparation of a diamino protected L-lysine represented by Formula (IV): wherein PG is an amine protecting group.
  • the diamino protected L-lysine represented by Formula (IV) is obtained by performing an amine protection of L-lysine monohydrochloride by using an amine protecting reagent in the presence of a suitable base and solvent.
  • the examples of the amine protecting group include, but are not limited to, t-butyloxycarbonyl (Boc), benzyloxycarbonyl (Cbz), allyloxycarbonyl (alloc), fluorenylmethyloxycarbonyl (Fmoc), trifluoroacetyl (TFA), acetyl (Ac), 2-(trimethylsilyl)ethoxycarbonyl (Teoc), tosyl (Ts), pivaloyl, and phthalimide.
  • the amino protecting group is benzyloxycarbonyl (Cbz), which may be from benzyl chloroformate as the amine protecting reagent.
  • the examples of the base for amine protection include, but are not limited to, N-methylmorpholine, diisopropylethyl amine (DIPEA), triethylamine (TEA), tri-n-propylamine, pyridine, alkaline metal hydroxides, and alkaline metal carbonates, such as sodium hydroxide, potassium hydroxide, calcium hydroxide, lithium hydroxide, sodium carbonate, potassium carbonate, calcium carbonate, sodium methoxide, potassium methoxide, sodium hydride, potassium hydride, and lithium hydride.
  • the base for amine protection is potassium hydroxide, potassium carbonate, or a combination thereof.
  • the examples of the solvent for amine protection include, but are not limited to, a polar protic solvent, such as methanol, ethanol, isopropyl alcohol, and water; or a polar aprotic solvent, such as dichloromethane, N-methylpyrrolidone, tetrahydrofuran, ethyl acetate, acetone, dimethylformamide, acetonitrile, and dimethyl sulfoxide; and a non-polar solvent, such as toluene, hexane, ether, and a mixture thereof.
  • the solvent for amine protection is water, methanol, or a combination thereof.
  • the reaction for preparing the diamino protected L-lysine represented by Formula (IV) may be carried out at a temperature of 0°C to 60°C for about 10 minutes to 24 hours.
  • the reaction may be carried out at about 10°C to 30°C in the presence of a base.
  • the completion of the reaction may be monitored by high pressure liquid chromatography or a suitable chromatographic technique.
  • the diamino protected L-lysine represented by Formula (IV) may be isolated by any standard method known in the art. In some embodiments, the resulting diamino protected L-lysine is isolated by filtration.
  • the method for preparing lisdexamphetamine or a salt thereof comprises coupling the diamino protected L-lysine represented by Formula (IV) and L-norephedrine represented by Formula (II) to obtain a compound represented by Formula (V) as illustrated below: wherein PG is an amine protecting group.
  • the coupling comprising converting the diamino protected L-lysine represented by Formula (IV) to an active intermediate by using an acid activating reagent in the presence of a base and a solvent.
  • the examples of the acid activating reagent include, but are not limited to, dicyclohexyl carbodiimide (DCC), 1-ethyl-3-(dimethylamino)carbodiimide hydrochloride (EDC-HC1), carbonyldiimidazole (CDI), benzotriazol-1-yloxytri(pyrrolidino)phosphonium hexafluorophosphate (PyBOP), (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate (HATU), O-(1H-benzotriazol-1-yl)-N,N,N,N-tetramethyluronium hexafluorophosphate (HBTU), N-hydroxybenzotriazole, N-hydroxysuccinimide (HOSu), and haloformate, such as methyl chloro
  • DCC dicyclohex
  • the equivalent of isobutyl chloroformate as the acid activating reagent may be 0.95 to 1.05. In some embodiments, the equivalent of isobutyl chloroformate is about 0.96, about 0.97, about 0.98, about 0.99, about 1.0, about 1.01, about 1.02, about 1.03, or about 1.04.
  • the examples of the base for acid activation include, but are not limited to, N-methylmorpholine, diisopropylethyl amine (DIPEA), triethylamine (TEA), tri-n-propylamine, pyridine, and 1,8-diazabicyclo(5.4.0)undec-7-ene (DBU).
  • the base for acid activation is N-methylmorpholine.
  • the equivalent of N-methylmorpholine as the base for acid activation may be 1.0 to 2.4. In some embodiments, the equivalent of N-methylmorpholine is about 1.1, about 1.3, about 1.5, about 1.7, about 1.9, about 2.0, about 2.1, or about 2.3.
  • the examples of the solvent for acid activation include, but are not limited to, a polar aprotic solvent, such as dichloromethane, N-methylpyrrolidone, tetrahydrofuran, ethyl acetate, acetone, dimethylformamide, acetonitrile, and dimethyl sulfoxide.
  • a polar aprotic solvent such as dichloromethane, N-methylpyrrolidone, tetrahydrofuran, ethyl acetate, acetone, dimethylformamide, acetonitrile, and dimethyl sulfoxide.
  • the solvent for acid activation is dichloromethane.
  • the coupling may be carried out by reacting L-norephedrine represented by Formula (II) and the diamino protected L-lysine represented by Formula (IV) with an acid activating reagent at a temperature of from -20°C to 40°C for about 10 minutes to 24 hours.
  • an acid activating reagent at a temperature of from -20°C to 40°C for about 10 minutes to 24 hours.
  • the reaction may be carried out at about -15°C to 30°C, such as - 15°C to 15°C, -15°C to 20°C, -10°C to 30°C, and -5°C to 25°C.
  • the completion of the reaction may be monitored by high pressure liquid chromatography or a suitable chromatographic technique.
  • the resulting compound represented by Formula (V) may be subjected to a subsequent step without further isolation.
  • the method for preparing lisdexamphetamine or a salt thereof comprises reacting the compound represented by Formula (V) with an acylation reagent to obtain the intermediate compound represented by Formula (VI) as illustrated below: wherein PG is an amine protecting group.
  • the intermediate compound represented by Formula (VI) is obtained through converting the hydroxyl group of the compound represented by Formula (V) to an O-acylation group by using an acylation reagent and a base.
  • the O-acylation group may be O-formyl, O-acetyl, O-trifluoroacetyl, O-benzoyl, O-acrylyl, or O-carbonate group.
  • the examples of the acylation reagent include, but are not limited to, formic acid, methyl formate, ethyl formate, acetic acid, acetic anhydride, ethyl acetate, acetyl chloride, trifluoroacetic acid, trifluoroacetic anhydride, benzoic acid, benzoic anhydride, benzoic methyl ester, benzoic chloride, acrylic acid, acryloyl chloride, di- tert -butyl dicarbonate, and aryl chloroformate.
  • the acylation reagent is acetic anhydride.
  • the equivalent of acetic anhydride as the acylating reagent may be 1.0 to 3.0. In some embodiments, the equivalent of acetic anhydride is about 1.2, about 1.4, about 1.5, about 1.6, about 1.8, about 2.0, about 2.2, about 2.4, about 2.5, about 2.6, or about 2.8.
  • the examples of the base for acylation include, but are not limited to, imidazole, guanidine, N-methylmorpholine, diisopropylethyl amine (DIPEA), triethylamine (TEA), tri-n-propylamine, pyridine, 2,6-lutidine, piperidine, pyrrole, pyrrolidine, and 1,8-diazabicyclo(5.4.0)undec-7-ene (DBU).
  • the base for acylation is pyridine.
  • the equivalent of pyridine as the base for acylation may be 1.0 to 3.0. In some embodiments, the equivalent of pyridine is about 1.2, about 1.4, about 1.5, about 1.6, about 1.8, about 2.0, about 2.2, about 2.4, about 2.5, about 2.6, or about 2.8.
  • the acylation reaction may be carried out at a temperature of 25°C to 100°C, such as 40°C to 100°C, for about 10 minutes to 24 hours.
  • the reaction may be carried out at about 60°C to about 90°C.
  • the completion of the reaction may be monitored by high pressure liquid chromatography or a suitable chromatographic technique.
  • the intermediate compound represented by Formula (VI) may be isolated by any standard method known in the art. In some embodiments, the resulting intermediate compound is isolated by filtration.
  • the method for preparing lisdexamphetamine or a salt thereof comprises reducing the O-acylation group of the intermediate compound represented by Formula (VI) through a catalytic hydrogenation to obtain lisdexamphetamine represented by Formula (I) as illustrated below: wherein PG is an amine protecting group.
  • the catalytic hydrogenation is carried out by contacting the intermediate compound represented by Formula (VI) by hydrogenation with a catalyst in the presence of a suitable solvent.
  • the catalyst for catalytic hydrogenation may be a transition metal catalyst, such as nickel, palladium, platinum, ruthenium, and rhodium.
  • the hydrogenation catalyst is Pd/C (palladium on charcoal).
  • the examples of the solvent for catalytic hydrogenation include, but are not limited to, a polar protic solvent, such as acetic acid, methanol, ethanol, isopropyl alcohol, and water; a polar aprotic solvent, such as dichloromethane, N-methylpyrrolidone, tetrahydrofuran, ethyl acetate, dimethylformamide, acetonitrile, and dimethyl sulfoxide; and a non-polar solvent, such as toluene, hexane, ether, and any combination thereof.
  • the solvent for catalytic hydrogenation comprises methanol and water.
  • the weight ratio of the intermediate compound represented by Formula (VI) to the solvent for catalytic hydrogenation may be from 1:2 to 1:20. In some embodiments, the weight ratio of the intermediate compound represented by Formula (VI) to the solvent for catalytic hydrogenation is about 1:5, about 1:10, about 1:12, about 1:15, or about 1:18.
  • the reduction reaction may be carried out at a temperature of 25°C to 80°C, such as 30°C to 40°C, for about 1 hour to 24 hours.
  • the completion of the reaction may be monitored by high pressure liquid chromatography or a suitable chromatographic technique.
  • the lisdexamphetamine represented by Formula (I) may be converted to its pharmaceutically acceptable salt by contacting with a corresponding acid thereof, such as methanesulfonic acid.
  • a corresponding acid thereof such as methanesulfonic acid.
  • the pharmaceutically acceptable salt of lisdexamfetamine may be isolated by filtration.
  • L-lysine monohydrochloride (91.3 g, 0.50 mole), potassium hydroxide (56.0 g, 0.50 mole), and potassium carbonate (152.0 g, 1.10 mole) were mixed in water (295.0 g) and methanol (500.0 g) at ambient temperature under inert atmosphere. The mixture was cooled to about 10°C, followed by adding benzyl chloroformate (187.6 g, 1.10 mole). After then, the reaction mixture was subjected to stirring at 25°C for a half hour. The reaction mixture was then filtered at 25°C and washed with methanol (230.0 g). The filtrate (1,273.0 g) was collected and concentrated at 60°C to obtain a residual (764.0 g).
  • the residual was extracted with toluene (250.0 g) and water (50.0 g).
  • the toluene layer was separated from the aqua layer, and then washed with water (150.0 g).
  • the toluene layer was abandoned, and the aqua layer was collected and acidified with 32% HC1 (91.0 g, 0.80 mole).
  • the acidified aqua was extracted with toluene (650.0 g), and then the toluene layer was separated from the aqua layer, and then washed twice with water (170.0 g and 100.0 g).
  • the toluene layer was concentrated under reduced pressure at 60°C to 80°C to form a crude diamino protected L-lysine (232.1 g) (in which the protection group (PG) was a carbobenzoxy (Cbz) group) with purity of 91.5%, which was measured by ultra-performance liquid chromatography, and with reaction yield of 95.6%.
  • the protection group (PG) was a carbobenzoxy (Cbz) group
  • the crude diamino protected L-lysine was further isolated by crystallization with ethyl acetate (460.0 g) and heptane (920.0 g) and filtration.
  • the wet cake of the diamino protected L-lysine represented by Formula (IV) (373.0 g) was obtained after filtration, and the dry cake (193.5 g) was obtained after drying.
  • the obtained diamino protected L-lysine represented by Formula (IV) had purity of 98.2%, measured by ultra-performance liquid chromatography, and yield of 93%.
  • Formula (IV) in which the PG was a Cbz group (165.6 g, 0.40 mole) as prepared in Example 1, and isobutyl chloroformate (55.0 g, 0.404 mole) were mixed in dichloromethane (501.0 g) at ambient temperature under inert atmosphere. The mixture was cooled to about -15°C, followed by adding N-methylmorpholine (84.5 g,
  • L-norephedrine represented by Formula (II) (60.4 g, 0.40 mole) was dissolved in dichloromethane (300.0 g) and slowly mixed with the reaction mixture containing the diamino protected L-lysine represented by Formula (IV), isobutyl chloroformate, and N-methylmorpholine.
  • reaction mixture was subjected to stirring for 1 hour at 0°C to 30°C, and then quenched with water (300.0 g).
  • the dichloromethane layer was separated from the aqua layer, and then washed with 5% HC1 (w/w) aqueous solution (100.0 g and 100.0 g) and water (100.0 g).
  • the dichloromethane layer was collected and concentrated under reduced pressure at 60°C to give a crude compound represented by Formula (V) (440.0 g) with the Cbz group as the PG.
  • Formula (V) was used to a subsequent step without further purification.
  • the compound represented by Formula (V) with the Cbz group as the PG as prepared in Example 2 (342.0 g) was mixed with acetic anhydride (62.0 g, 0.61 mole) and pyridine (49.0 g, 0.62 mole), followed by stirring at ambient temperature under inert atmosphere. The mixture was heated to about 80°C to 90°C and subjected to stirring for seven hours. After stirring for seven hours, the reaction mixture was concentrated under reduced pressure to obtain about 370 g of a residual.
  • the crude wet cake of the compound represented by Formula (VI) (325.0) was mixed in ethyl acetate (1,480.0 g) and heated to reflux (about 77°C) to form a homogenous solution. The solution was cooled down slowly to about 1°C to 2°C and hold for another 2 hours.
  • the wet cake of the compound represented by Formula (VI) (280.0 g) (in which PG was the Cbz group and R 1 was methyl) was obtained after filtration.
  • the compound represented by Formula (VI) in white dry cake (170.0 g) was obtained with purity of 99.79%, measured by ultra-performance liquid chromatography, and yield of 72% (2 steps).
  • the oily compound represented by Formula (I) (4.5 g) as prepared in Example 4 was dissolved in isopropyl alcohol (45.0 g) and stirred at ambient temperature under inert atmosphere. To the reaction mixture, methanesulfonic acid (3.2 g) was slowly added. Further, the reaction mixture was heated to 55°C to 65°C and stirred for one hour. The reaction mixture was then cooled down slowly to room temperature (about 25°C), stirred for 3 hours, filtered, washed with isopropyl alcohol (22.5 g) and dried under vacuum to obtain 5.5 g of lisdexamfetamine dimesylate with purity of 99.9%, measured by ultra-performance liquid chromatography.
  • the compound represented by Formula (IX) was mixed and stirred with heptane (795 g), water (1,295 g) and sodium hydroxide (80.0 g, 2.0 mole) at ambient temperature under inert atmosphere until all solid dissolved.
  • the heptane layer was separated from the aqua layer, and washed with water (100.0 g).
  • the S-form of the compound of Formula (VIII) was generated in the heptane layer, and then mixed with isopropyl alcohol (200.0 g).
  • 5% Palladium on charcoal (10 g) was added to the reaction mixture and kept under 0.4 to 0.5 MPa hydrogen pressure for 4 hours at 30°C to 35°C, and for 4 hours at 60°C to 65°C.
  • Phenylpropanolamine (PPA) (302.0 g, 2.0 mole) was mixed with isopropyl alcohol (IPA) (698.0 g) at 50°C to form a clear solution under inert atmosphere.
  • IPA isopropyl alcohol
  • (+)-Dibenzoyl-D-tartaric acid (D-DBTA) (178.6 g, 0.48 mole) was dissolved in IPA (774.1 g) and slowly charged into PPA/IPA solution over three hours at 25°C to 50°C. The solid was formed during charging. After charging, the reaction mixture was subjected to stirring at about 25°C for two hours. After filtration, the crude wet cake (346.5 g) of the compound represented by Formula (X) was obtained with enantiomeric excess (e.e.) purity of about 95.7%, measured by chiral liquid chromatography.
  • the crude wet cake of the compound represented by Formula (X) (346.5) was dissolved in methanol (1,200 g) and heated to reflux (about 66°C) to distill out methanol (609.0 g) under inert atmosphere. The mixture solution was cooled down slowly to about 1°C to 2°C and hold for another two hours.
  • the wet cake of the compound represented by Formula (X) was obtained after filtration, and the dry cake (258.5 g) of the compound represented by Formula (X) was obtained after drying (e.e. purity of about 99.99%, measured by chiral liquid chromatography).
  • the compound represented by Formula (X) (132.0 g, 0.4 mole) was mixed and stirred with cyclopentyl methyl ether (CPME) (250.0 g) and 2 M HCl (aq) (400.0 g) at 75°C under inert atmosphere until all solid dissolved.
  • the CPME layer was separated from the aqua layer, and washed with 1 M HCl (aq) (100.0 g).
  • L-norephedrine of Formula (II) was generated in the collected HCl (aq) layer, and then neutralized with 45% NaOH (82.5 g, 0.93 mole) and extracted with dichloromethane four times (213.0 g, 103.0 g, 100.0 g and 100.0 g, respectively).
  • the collected dichloromethane layer was concentrated under reduced pressure to obtain the residual (66.0 g) of L-norephedrine, in which the yield was 38.0%, from phenylpropanolamine.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

Provided is a compound represented by Formula (VI) for preparing lisdexamphetamine or a salt thereof. Also provided is a method for preparing lisdexamfetamine or a salt thereof including performing reduction and debenzylation of the compound represented by Formula (VI) by hydrogenation.

Description

    BACKGROUND 1. Technical Field
  • The present disclosure relates to compounds for preparing lisdexamfetamine, and to methods for preparing lisdexamfetamine from the compounds.
  • 2. Description of Associated Art
  • Lisdexamfetamine dimesylate, i.e., (2S)-2,6-diamino-N-[(1S)-1-methyl-2-phenylethyl] hexanamide dimethanesulfonate under the brand name of Vyvanse, is used to treat attention deficit hyperactivity disorder (ADHD) in adults and over 6-year-old children. Also, it was approved to treat moderate to severe binge eating disorder (BED) by the US FDA in 2015. Lisdexamfetamine dimesylate increases attention and decreases restlessness in children and adults who are overactive, unable to concentrate for long periods of time, or easily distracted and impulsive. This medicament is usually used as a part of a total treatment program that includes social, educational, and psychological treatments.
    Figure imgb0001
  • As to the preparation of lisdexamfetamine dimesylate, U.S. Patent Publication No. 2012/0157706 discloses a method for preparation of lisdexamfetamine dimesylate from D-amphetamine. The method involves reacting D-amphetamine with (S)-2,5-dioxopyrrolidin-1-yl 2,6-bis(benzyloxycarbonylamino) hexanoate to form a lysine amphetamine bearing benzylcarbamate protecting groups. The lysine amphetamine bearing the benzylcarbamate protecting groups is then converted to lisdexamfetamine dimesylate by catalytic hydrogenation to remove the benzylcarbamate protecting groups, followed by subsequent addition of methanesulfonic acid to generate the lisdexamfetamine dimesylate.
  • International Patent Publication No. WO 2010/042120 discloses a process for preparation of lisdexamfetamine by reacting D-amphetamine with (S)-2,6-bis((tert-butoxycarbonyl)amino)hexanoic acid in the presence of an alkylphosphonic acid anhydride (e.g., T3P) as a coupling agent under a base condition including a solvent, followed by adding methanesulfonic acid to remove the tert-butoxycarbonyl protecting groups and thus generate lisdexamfetamine dimesylate. In addition, International Patent Publication No. WO 2013/011526 discloses another process for preparation of lisdexamfetamine by reacting D-amphetamine with (S)-2,6-bis((tert-butoxycarbonyl)amino)hexanoic acid in the presence of isobutyl chloroformate as a coupling agent under a base condition including a solvent.
  • International Patent Publication No. WO 2010/148305 discloses a process for preparation of lisdexamfetamine starting from L-norephedrine. First, L-norephedrine is converted to the corresponding chloro-D-amphetamine hydrochloride by thionyl chloride, and the resulting chloro-D-amphetamine hydrochloride is coupled with (S)-2,6-bis(2,2,2-trifluoroacetamido)hexanoic acid to obtain an N,N'-bis-trifluoroacetyl-protected chloro-lisdexamfetamine intermediate. Further, the N,N'-bis-trifluoroacetyl-protected chloro-lisdexamfetamine intermediate is converted to N,N'-bis-trifluoroacetyl-protected lisdexamfetamine by removal of chlorine with catalytic hydrogenation. Subsequently, lisdexamfetamine dimesylate is obtained by subjecting N,N'-bis-trifluoroacetyl-protected lisdexamfetamine to a hydrolysis reaction by methanesulfonic acid.
  • U.S. Patent Publication No. 2016/0376618 discloses a method for preparation of lisdexamfetamine from amfetamine, comprising reacting amfetamine with N,N'-bis-acyllysine ester and an enzyme catalyst to form a lisdexamfetamine stereoisomer, followed by converting the stereoisomer to lisdexamfetamine by treating with tetrakis(triphenylphosphine)palladium to remove an allyloxycarbonyl protecting group. Subsequently, by addition of methanesulfonic acid, lisdexamfetamine dimesylate is obtained.
  • International Patent Publication No. WO 2017/098533 discloses a process for preparation of lisdexamfetamine from (2S,3R)-2-methyl-3-phenylaziridine. In the disclosed process, (2S,3R)-2-methyl-3-phenylaziridine is coupled with Lys-2Boc-NHS to form a corresponding aziridine compound. The aziridine compound is then converted to N,N'-bis-Boc-protected lisdexamfetamine by ring-opening with catalytic hydrogenation, and further converted to lisdexamfetamine dimesylate by addition of methanesulfonic acid to decarboxylate the tert-butoxycarbonyl protecting groups.
  • The procedures described in above literatures mostly employ D-amphetamine as a starting material. D-amphetamine, also known as dextroamphetamine, is a Schedule II drug defined by the United States Controlled Substances Act. Therefore, special licenses, handling procedures, and compliance with governmental regulatory provisions are required for using large quantities of the controlled substance. As to other preparation processes, chloro-D-amphetamine or phenylaziridine is used as a starting material to replace the D-amphetamine. However, these processes involve additional steps of inserting a chloro group and thereafter removing the chloro group. In addition, multi-steps are required for synthesis of phenylaziridine.
  • Accordingly, there is still an unmet need for safe and effective methods and intermediates for preparing lisdexamfetamine and its pharmaceutically acceptable salts without using D-amphetamine as a starting material.
  • SUMMARY
  • The present disclosure provides a method for preparing lisdexamfetamine represented by Formula (I) below and a salt thereof:
    Figure imgb0002
  • Also, the present disclosure provides a compound represented by Formula (VI) below or a salt thereof for preparing lisdexamphetamine or a salt thereof:
    Figure imgb0003
    wherein R1 is hydrogen, an aliphatic group, C1-C6 alkoxyl, a C1-C6 alkylamino group, or an aromatic group, and PG is an amine protecting group.
  • In at least one embodiment of the present disclosure, in the compound represented by Formula (VI), R1 is methyl. In some embodiments, PG is benzyloxycarbonyl. In some embodiments, the compound for preparing lisdexamphetamine or a salt thereof is represented by Formula (VIa) below:
    Figure imgb0004
    wherein Cbz refers to benzyloxycarbonyl.
  • In at least one embodiment of the present disclosure, the method for preparing lisdexamfetamine or a salt thereof provided herein comprises reducing an O-acylation group of an intermediate compound represented by Formula (VI) or a salt thereof through a catalytic hydrogenation to obtain the lisdexamphetamine or the salt thereof:
    Figure imgb0005
    wherein R1 is hydrogen, an aliphatic group, C1-C6 alkoxyl, a C1-C6 alkylamino group, or an aromatic group, and PG is an amine protecting group.
  • In at least one embodiment of the present disclosure, as illustrated below, the method further comprises reacting a compound represented by Formula (V) or a salt thereof with an acylation reagent to obtain the intermediate compound represented by Formula (VI):
    Figure imgb0006
    wherein R1 and PG are defined as above.
  • In at least one embodiment of the present disclosure, as illustrated below, the method further comprises coupling L-norephedrine represented by Formula (II) or a salt thereof and a diamino protected L-lysine represented by Formula (IV) or a salt thereof to obtain the compound represented by Formula (V):
    Figure imgb0007
    wherein PG is defined as above.
  • In some embodiments of the present disclosure, as illustrated below, the coupling L-norephedrine or the salt thereof and the diamino protected L-lysine or the salt thereof is carried out by reaction with an acid activating reagent:
    Figure imgb0008
    wherein PG is defined as above.
  • In at least one embodiment of the present disclosure, as illustrated below, the method further comprises reacting L-lysine represented by Formula (III) below with an amine protecting group in the presence of a protection reagent and a solvent to obtain the diamino protected L-lysine represented by Formula (IV):
    Figure imgb0009
    wherein PG is the amine protecting group.
  • In at least one embodiment of the present disclosure, the method further comprises contacting the lisdexamphetamine with methanesulfonic acid to obtain lisdexamfetamine dimesylate.
  • DETAILED DESCRIPTION OF THE EMBODIMENTS
  • The following examples are used to exemplify the present disclosure. A person of ordinary skill in the art can conceive the other advantages and effects of the present disclosure, based on the disclosure of the specification. The present disclosure can also be implemented or applied as described in different examples. It is possible to modify and/or alter the examples for carrying out this disclosure without contravening its scope, for different aspects and applications.
  • It is further noted that, as used in this disclosure, the singular forms "a," "an," and "the" include plural referents unless expressly and unequivocally limited to one referent. The term "or" is used interchangeably with the term "and/or" unless the context clearly indicates otherwise.
  • As used herein, the term "comprising" or "comprises" is used in reference to compositions, methods, and respective component(s) thereof, which are included in the present disclosure, yet open to the inclusion of unspecified elements. For example, a composition, mixture, process or method that comprises a list of elements or actions is not necessarily limited to only those elements or actions, but may include other elements or actions not expressly listed, or inherent to such composition, mixture, process, or method.
  • As used herein, the terms "first," "second," "third," and the like (if present) are used to distinguish similar objects and are not necessarily used to describe a particular order. It should be understood that the objects are interchangeable at an appropriate time.
  • As used herein, the term "about" generally means within 10%, 5%, 1%, or 0.5% of a given value or range. Alternatively, the term "about" means within an acceptable standard error of the mean when considered by one of ordinary skill in the art. Unless otherwise expressly specified, all of the numerical ranges, amounts, values and percentages such as those for quantities of materials, durations of time periods, temperatures, operating conditions, ratios of amounts, and the likes disclosed herein should be understood as modified in all instances by the term "about."
  • The present disclosure is directed to an intermediate compound and a method for preparing lisdexamphetamine or a salt thereof. In at least one embodiment of the present disclosure, the intermediate compound for the preparation method is represented by Formula (VI) below:
    Figure imgb0010
    or a salt thereof, wherein R1 is hydrogen, an aliphatic group, C1-C6 alkoxyl, a C1-C6 alkylamino group, or an aromatic group, and PG is an amine protecting group.
  • In at least one embodiment of the present disclosure, an exemplary process for preparing lisdexamfetamine dimesylate by the method provided herein is shown in Scheme 1 below:
    Figure imgb0011
  • In the Scheme 1 illustrated above, R1 is hydrogen, an aliphatic group, C1-C6 alkoxyl, a C1-C6 alkylamino, or an aromatic group, and PG is an amine protecting group.
  • In at least one embodiment of the present disclosure, the method for preparing lisdexamfetamine represented by Formula (I) and a salt thereof may start from preparation of a diamino protected L-lysine represented by Formula (IV):
    Figure imgb0012
    wherein PG is an amine protecting group.
  • In some embodiments, the diamino protected L-lysine represented by Formula (IV) is obtained by performing an amine protection of L-lysine monohydrochloride by using an amine protecting reagent in the presence of a suitable base and solvent.
  • In at least one embodiment of the present disclosure, the examples of the amine protecting group include, but are not limited to, t-butyloxycarbonyl (Boc), benzyloxycarbonyl (Cbz), allyloxycarbonyl (alloc), fluorenylmethyloxycarbonyl (Fmoc), trifluoroacetyl (TFA), acetyl (Ac), 2-(trimethylsilyl)ethoxycarbonyl (Teoc), tosyl (Ts), pivaloyl, and phthalimide. In some embodiments, the amino protecting group is benzyloxycarbonyl (Cbz), which may be from benzyl chloroformate as the amine protecting reagent.
  • In at least one embodiment of the present disclosure, the examples of the base for amine protection include, but are not limited to, N-methylmorpholine, diisopropylethyl amine (DIPEA), triethylamine (TEA), tri-n-propylamine, pyridine, alkaline metal hydroxides, and alkaline metal carbonates, such as sodium hydroxide, potassium hydroxide, calcium hydroxide, lithium hydroxide, sodium carbonate, potassium carbonate, calcium carbonate, sodium methoxide, potassium methoxide, sodium hydride, potassium hydride, and lithium hydride. In some embodiments, the base for amine protection is potassium hydroxide, potassium carbonate, or a combination thereof.
  • In at least one embodiment of the present disclosure, the examples of the solvent for amine protection include, but are not limited to, a polar protic solvent, such as methanol, ethanol, isopropyl alcohol, and water; or a polar aprotic solvent, such as dichloromethane, N-methylpyrrolidone, tetrahydrofuran, ethyl acetate, acetone, dimethylformamide, acetonitrile, and dimethyl sulfoxide; and a non-polar solvent, such as toluene, hexane, ether, and a mixture thereof. In some embodiments, the solvent for amine protection is water, methanol, or a combination thereof.
  • In at least one embodiment of the present disclosure, the reaction for preparing the diamino protected L-lysine represented by Formula (IV) may be carried out at a temperature of 0°C to 60°C for about 10 minutes to 24 hours. When using a benzyloxycarbonyl group as an amine protecting group, the reaction may be carried out at about 10°C to 30°C in the presence of a base. The completion of the reaction may be monitored by high pressure liquid chromatography or a suitable chromatographic technique. The diamino protected L-lysine represented by Formula (IV) may be isolated by any standard method known in the art. In some embodiments, the resulting diamino protected L-lysine is isolated by filtration.
  • In at least one embodiment of the present disclosure, the method for preparing lisdexamphetamine or a salt thereof comprises coupling the diamino protected L-lysine represented by Formula (IV) and L-norephedrine represented by Formula (II) to obtain a compound represented by Formula (V) as illustrated below:
    Figure imgb0013
    wherein PG is an amine protecting group.
  • In at least one embodiment of the present disclosure, the coupling comprising converting the diamino protected L-lysine represented by Formula (IV) to an active intermediate by using an acid activating reagent in the presence of a base and a solvent.
  • In at least one embodiment of the present disclosure, the examples of the acid activating reagent include, but are not limited to, dicyclohexyl carbodiimide (DCC), 1-ethyl-3-(dimethylamino)carbodiimide hydrochloride (EDC-HC1), carbonyldiimidazole (CDI), benzotriazol-1-yloxytri(pyrrolidino)phosphonium hexafluorophosphate (PyBOP), (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate (HATU), O-(1H-benzotriazol-1-yl)-N,N,N,N-tetramethyluronium hexafluorophosphate (HBTU), N-hydroxybenzotriazole, N-hydroxysuccinimide (HOSu), and haloformate, such as methyl chloroformate, ethyl chloroformate, isopropyl chloroformate, isobutyl chloroformate, benzyl chloroformate, phenyl chloroformate, and aryloxy chloroformate. In some embodiments, the acid activating reagent is isobutyl chloroformate.
  • In at least one embodiment of the present disclosure, the equivalent of isobutyl chloroformate as the acid activating reagent may be 0.95 to 1.05. In some embodiments, the equivalent of isobutyl chloroformate is about 0.96, about 0.97, about 0.98, about 0.99, about 1.0, about 1.01, about 1.02, about 1.03, or about 1.04.
  • In at least one embodiment of the present disclosure, the examples of the base for acid activation include, but are not limited to, N-methylmorpholine, diisopropylethyl amine (DIPEA), triethylamine (TEA), tri-n-propylamine, pyridine, and 1,8-diazabicyclo(5.4.0)undec-7-ene (DBU). In some embodiments, the base for acid activation is N-methylmorpholine.
  • In at least one embodiment of the present disclosure, the equivalent of N-methylmorpholine as the base for acid activation may be 1.0 to 2.4. In some embodiments, the equivalent of N-methylmorpholine is about 1.1, about 1.3, about 1.5, about 1.7, about 1.9, about 2.0, about 2.1, or about 2.3.
  • In at least one embodiment of the present disclosure, the examples of the solvent for acid activation include, but are not limited to, a polar aprotic solvent, such as dichloromethane, N-methylpyrrolidone, tetrahydrofuran, ethyl acetate, acetone, dimethylformamide, acetonitrile, and dimethyl sulfoxide. In some embodiments, the solvent for acid activation is dichloromethane.
  • In at least one embodiment of the present disclosure, the coupling may be carried out by reacting L-norephedrine represented by Formula (II) and the diamino protected L-lysine represented by Formula (IV) with an acid activating reagent at a temperature of from -20°C to 40°C for about 10 minutes to 24 hours. When using isobutyl chloroformate as an acid activating reagent, the reaction may be carried out at about -15°C to 30°C, such as - 15°C to 15°C, -15°C to 20°C, -10°C to 30°C, and -5°C to 25°C. The completion of the reaction may be monitored by high pressure liquid chromatography or a suitable chromatographic technique. The resulting compound represented by Formula (V) may be subjected to a subsequent step without further isolation.
  • In at least one embodiment of the present disclosure, the method for preparing lisdexamphetamine or a salt thereof comprises reacting the compound represented by Formula (V) with an acylation reagent to obtain the intermediate compound represented by Formula (VI) as illustrated below:
    Figure imgb0014
    wherein PG is an amine protecting group.
  • In at least one embodiment of the present disclosure, the intermediate compound represented by Formula (VI) is obtained through converting the hydroxyl group of the compound represented by Formula (V) to an O-acylation group by using an acylation reagent and a base.
  • In at least one embodiment of the present disclosure, the O-acylation group may be O-formyl, O-acetyl, O-trifluoroacetyl, O-benzoyl, O-acrylyl, or O-carbonate group. In some embodiments, the examples of the acylation reagent include, but are not limited to, formic acid, methyl formate, ethyl formate, acetic acid, acetic anhydride, ethyl acetate, acetyl chloride, trifluoroacetic acid, trifluoroacetic anhydride, benzoic acid, benzoic anhydride, benzoic methyl ester, benzoic chloride, acrylic acid, acryloyl chloride, di-tert-butyl dicarbonate, and aryl chloroformate. In some embodiments, the acylation reagent is acetic anhydride.
  • In at least one embodiment of the present disclosure, the equivalent of acetic anhydride as the acylating reagent may be 1.0 to 3.0. In some embodiments, the equivalent of acetic anhydride is about 1.2, about 1.4, about 1.5, about 1.6, about 1.8, about 2.0, about 2.2, about 2.4, about 2.5, about 2.6, or about 2.8.
  • In at least one embodiment of the present disclosure, the examples of the base for acylation include, but are not limited to, imidazole, guanidine, N-methylmorpholine, diisopropylethyl amine (DIPEA), triethylamine (TEA), tri-n-propylamine, pyridine, 2,6-lutidine, piperidine, pyrrole, pyrrolidine, and 1,8-diazabicyclo(5.4.0)undec-7-ene (DBU). In some embodiments, the base for acylation is pyridine.
  • In at least one embodiment of the present disclosure, the equivalent of pyridine as the base for acylation may be 1.0 to 3.0. In some embodiments, the equivalent of pyridine is about 1.2, about 1.4, about 1.5, about 1.6, about 1.8, about 2.0, about 2.2, about 2.4, about 2.5, about 2.6, or about 2.8.
  • In at least one embodiment of the present disclosure, the acylation reaction may be carried out at a temperature of 25°C to 100°C, such as 40°C to 100°C, for about 10 minutes to 24 hours. When performing O-acetate reaction by acetic anhydride, the reaction may be carried out at about 60°C to about 90°C. The completion of the reaction may be monitored by high pressure liquid chromatography or a suitable chromatographic technique. The intermediate compound represented by Formula (VI) may be isolated by any standard method known in the art. In some embodiments, the resulting intermediate compound is isolated by filtration.
  • In at least one embodiment of the present disclosure, the method for preparing lisdexamphetamine or a salt thereof comprises reducing the O-acylation group of the intermediate compound represented by Formula (VI) through a catalytic hydrogenation to obtain lisdexamphetamine represented by Formula (I) as illustrated below:
    Figure imgb0015
    wherein PG is an amine protecting group.
  • In at least one embodiment of the present disclosure, the catalytic hydrogenation is carried out by contacting the intermediate compound represented by Formula (VI) by hydrogenation with a catalyst in the presence of a suitable solvent.
  • In at least one embodiment of the present disclosure, the catalyst for catalytic hydrogenation may be a transition metal catalyst, such as nickel, palladium, platinum, ruthenium, and rhodium. In some embodiments, the hydrogenation catalyst is Pd/C (palladium on charcoal).
  • In at least one embodiment of the present disclosure, the examples of the solvent for catalytic hydrogenation include, but are not limited to, a polar protic solvent, such as acetic acid, methanol, ethanol, isopropyl alcohol, and water; a polar aprotic solvent, such as dichloromethane, N-methylpyrrolidone, tetrahydrofuran, ethyl acetate, dimethylformamide, acetonitrile, and dimethyl sulfoxide; and a non-polar solvent, such as toluene, hexane, ether, and any combination thereof. In some embodiments, the solvent for catalytic hydrogenation comprises methanol and water.
  • In at least one embodiment of the present disclosure, the weight ratio of the intermediate compound represented by Formula (VI) to the solvent for catalytic hydrogenation may be from 1:2 to 1:20. In some embodiments, the weight ratio of the intermediate compound represented by Formula (VI) to the solvent for catalytic hydrogenation is about 1:5, about 1:10, about 1:12, about 1:15, or about 1:18.
  • In at least one embodiment of the present disclosure, the reduction reaction may be carried out at a temperature of 25°C to 80°C, such as 30°C to 40°C, for about 1 hour to 24 hours. The completion of the reaction may be monitored by high pressure liquid chromatography or a suitable chromatographic technique.
  • In at least one embodiment of the present disclosure, the lisdexamphetamine represented by Formula (I) may be converted to its pharmaceutically acceptable salt by contacting with a corresponding acid thereof, such as methanesulfonic acid. In some embodiments, the pharmaceutically acceptable salt of lisdexamfetamine may be isolated by filtration.
  • Many examples have been used to illustrate the present disclosure. The examples below should not be taken as a limit to the scope of the present disclosure.
  • EXAMPLES Example 1: Preparation of the diamino protected L-lysine represented by Formula (IV)
  • Figure imgb0016
  • L-lysine monohydrochloride (91.3 g, 0.50 mole), potassium hydroxide (56.0 g, 0.50 mole), and potassium carbonate (152.0 g, 1.10 mole) were mixed in water (295.0 g) and methanol (500.0 g) at ambient temperature under inert atmosphere. The mixture was cooled to about 10°C, followed by adding benzyl chloroformate (187.6 g, 1.10 mole). After then, the reaction mixture was subjected to stirring at 25°C for a half hour. The reaction mixture was then filtered at 25°C and washed with methanol (230.0 g). The filtrate (1,273.0 g) was collected and concentrated at 60°C to obtain a residual (764.0 g).
  • Further, the residual was extracted with toluene (250.0 g) and water (50.0 g). The toluene layer was separated from the aqua layer, and then washed with water (150.0 g). The toluene layer was abandoned, and the aqua layer was collected and acidified with 32% HC1 (91.0 g, 0.80 mole). The acidified aqua was extracted with toluene (650.0 g), and then the toluene layer was separated from the aqua layer, and then washed twice with water (170.0 g and 100.0 g). The toluene layer was concentrated under reduced pressure at 60°C to 80°C to form a crude diamino protected L-lysine (232.1 g) (in which the protection group (PG) was a carbobenzoxy (Cbz) group) with purity of 91.5%, which was measured by ultra-performance liquid chromatography, and with reaction yield of 95.6%.
  • The crude diamino protected L-lysine was further isolated by crystallization with ethyl acetate (460.0 g) and heptane (920.0 g) and filtration. The wet cake of the diamino protected L-lysine represented by Formula (IV) (373.0 g) was obtained after filtration, and the dry cake (193.5 g) was obtained after drying. The obtained diamino protected L-lysine represented by Formula (IV) had purity of 98.2%, measured by ultra-performance liquid chromatography, and yield of 93%.
  • Example 2: Preparation of the compound represented by Formula (V)
  • Figure imgb0017
  • The diamino protected L-lysine represented by Formula (IV), in which the PG was a Cbz group (165.6 g, 0.40 mole) as prepared in Example 1, and isobutyl chloroformate (55.0 g, 0.404 mole) were mixed in dichloromethane (501.0 g) at ambient temperature under inert atmosphere. The mixture was cooled to about -15°C, followed by adding N-methylmorpholine (84.5 g, 0.84 mole). After addition of N-methylmorpholine, the reaction mixture was subjected to stirring at -10°C to 0°C for a half hour. L-norephedrine represented by Formula (II) (60.4 g, 0.40 mole) was dissolved in dichloromethane (300.0 g) and slowly mixed with the reaction mixture containing the diamino protected L-lysine represented by Formula (IV), isobutyl chloroformate, and N-methylmorpholine.
  • Further, the reaction mixture was subjected to stirring for 1 hour at 0°C to 30°C, and then quenched with water (300.0 g). The dichloromethane layer was separated from the aqua layer, and then washed with 5% HC1 (w/w) aqueous solution (100.0 g and 100.0 g) and water (100.0 g). The dichloromethane layer was collected and concentrated under reduced pressure at 60°C to give a crude compound represented by Formula (V) (440.0 g) with the Cbz group as the PG.
  • The crude compound represented by Formula (V) was dissolved with AcOH (100.0 g) at 70°C and concentrated to obtain 342 g of a residual with 93.3% of purity, measured by ultra-performance liquid chromatography. The residual of the compound represented by
  • Formula (V) was used to a subsequent step without further purification.
  • Example 3: Preparation of the compound represented by Formula (VI)
  • Figure imgb0018
  • The compound represented by Formula (V) with the Cbz group as the PG as prepared in Example 2 (342.0 g) was mixed with acetic anhydride (62.0 g, 0.61 mole) and pyridine (49.0 g, 0.62 mole), followed by stirring at ambient temperature under inert atmosphere. The mixture was heated to about 80°C to 90°C and subjected to stirring for seven hours. After stirring for seven hours, the reaction mixture was concentrated under reduced pressure to obtain about 370 g of a residual.
  • Ethyl acetate (980.0 g) was added to the residual and heated (75°C) to form a homogenous solution. The solution was cooled down slowly to about 1°C to 2°C and hold for another 2 hours. After filtration, the crude wet cake of the compound represented by Formula (VI) (325.0 g) (in which PG was the Cbz group and R1 was methyl) was obtained with purity of 98.9% measured by ultra-performance liquid chromatography.
  • For recrystallization, the crude wet cake of the compound represented by Formula (VI) (325.0) was mixed in ethyl acetate (1,480.0 g) and heated to reflux (about 77°C) to form a homogenous solution. The solution was cooled down slowly to about 1°C to 2°C and hold for another 2 hours. The wet cake of the compound represented by Formula (VI) (280.0 g) (in which PG was the Cbz group and R1 was methyl) was obtained after filtration. After drying, the compound represented by Formula (VI) in white dry cake (170.0 g) was obtained with purity of 99.79%, measured by ultra-performance liquid chromatography, and yield of 72% (2 steps).
    Figure imgb0019
  • In an embodiment, the compound was represented by Formula (VIa) above: m.p. = 166~167°C. IR (cm-1) 3324 (br), 3288, 3089 (w), 3063 (w), 3033 (w), 2939, 2856, 1736, 1687, 1649, 1536, 1497, 1454, 1371, 1302, 1244, 1233, 1137, 1082, 1041, 977, 913, 754, 698. 1H NMR (600 MHz, DMSO-d6) δ (ppm) 0.95 (d, 3H), 1.11-1.17 (m, 2H), 1.30-1.37 (m, 4H), 2.08 (s, 3H), 2.95 (dt, 2H), 3.91 (dt, 1H), 4.20 (m, 1H), 5.02 (s, 2H, 2H), 5.71 (d, 1H), 7.21 (t, 1H), 7.23 (d, 1H), 7.27-7.37 (Ar, 15H), 7.94 (d, 1H). 13C NMR (150 MHz, DMSO-d6) δ (ppm) 15.00 (CH3), 20.76 (CH3), 22.47 (CH2), 29.13 (CH2), 31.54 (CH2), 40.06 (CH2), 47.83 (CH), 54.42 (CH), 65.13 (CH2), 65.34 (CH2), 76.33 (CH), 126.46 (CH), 127.12 (CH), 127.66 (CH), 127.72 (CH), 128.15 (CH), 128.32 (CH), 137.06 (C), 137.28 (C), 137.97 (C), 155.87 (C), 156.07 (C), 169.68 (C), 171.21 (C). MS (EI): m/z 590 (M)+.
  • The performance of the compound represented by Formula (VI) was determined by using ultra-performance liquid chromatography. The conditions of ultra-performance liquid chromatography assay were described in Table 1 below. Table 1
    UV detector Waters Model code UPL Serial # C15UPL 183A
    Auto s ampler Waters Model code SDI Serial # C15SDI 107G
    Pump Waters Model code QSM Serial # C15QSM 379A
    Column tvpe Waters ACQUITY UPLC BEH C8, 2.1(ID) × 50 mm, 1.7 µm
    Detector 220 nm
    Column temperature 35°C
    Sample temperature 25°C
    Run time 8 min
    Dilute solvent MeOH
    Injection volume 1 µL
    UPLC Gradient Pump Program
       Mobile A 1,000 mL water / 50 mL acetonitrile / 1 g TFA
       Mobile B Acetonitrile
    Gradient A% B% Flow rate
       0 min 100 0 0.6 mL/min
       1 min 100 0 0.6 mL/min
       6 min 20 80 0.6 mL/min
       7 min 0 100 0.8 mL/min
       7.1 min 100 0 0.6 mL/min
       8 min 100 0 0.6 mL/min
  • Example 4: Preparation of lisdexamphetamine from the compound represented by Formula (VI)
  • Figure imgb0020
  • The compound represented by Formula (VI) with PG being the Cbz group and R1 being methyl (10.0 g, 16.98 mole) as prepared in Example 3 was mixed with methanol (80.0 g) and water (20.0 g). 5% Palladium on charcoal (0.50 g) was added to the reaction mixture and kept under 0.4 to 0.5 MPa hydrogen pressure for 16 hours at 30°C to 35°C. The completion of reaction was checked by liquid chromatography. Palladium on charcoal was filtered and washed with methanol and water combined solvent (20.0 g and 5.0g, respectively). The reaction filtrate was then concentrated under reduced pressure at 50°C to 80°C. The oily residual (4.5 g) of lisdexamphetamine represented by Formula (I) was obtained with purity of 99.2%, measured by ultra-performance liquid chromatography, and used to a subsequent step without further purification.
  • Example 5: Preparation of lisdexamphetamine dimesylate from lisdexamphetamine
  • Figure imgb0021
  • The oily compound represented by Formula (I) (4.5 g) as prepared in Example 4 was dissolved in isopropyl alcohol (45.0 g) and stirred at ambient temperature under inert atmosphere. To the reaction mixture, methanesulfonic acid (3.2 g) was slowly added. Further, the reaction mixture was heated to 55°C to 65°C and stirred for one hour. The reaction mixture was then cooled down slowly to room temperature (about 25°C), stirred for 3 hours, filtered, washed with isopropyl alcohol (22.5 g) and dried under vacuum to obtain 5.5 g of lisdexamfetamine dimesylate with purity of 99.9%, measured by ultra-performance liquid chromatography.
  • Example 6: Preparation of L-norephedrine from propiophenone
  • Figure imgb0022
  • Propiophenone (268.0 g, 2.0 mole) was mixed with Br2 (3.2 g, 0.02 mole) at 30°C under inert atmosphere. Until the initial red color disappeared, N-bromosuccinimide (356.0 g, 2.0 mole) was added into the reaction mixture at the range of 35°C to 45°C, and then acetic acid (26.8 g, 0.45 mole) was added. After adding, the reaction mixture was subjected to stirring at about 35°C for one hour. The completion of reaction was checked by liquid chromatography. The reaction mixture was extracted with heptane (450.0 g) and water (870.0 g). The heptane layer was separated from the aqua layer, and washed twice with water (100.0 g and 100.0 g). The compound represented by Formula (VII) was produced in the heptane layer, and used for the next step without further purification.
  • Potassium carbonate (828.0 g, 6 eq) was dissolved in water (972.0 g) to form a clear solution (1,800.0 g), which was then mixed with benzyl amine (220.0 g, 2.05 mole). The BnNH2/K2CO3/water mixture was cooled to about 10°C, followed by adding the compound represented by Formula (VII)/heptane layer. After adding, the reaction mixture was subjected to stirring at about 30°C for 16 hours. The heptane layer was separated from the aqua layer, and washed twice with water (100.0 g and 100.0 g). The heptane layer was concentrated under reduced pressure at about 60°C to obtain a residual (500.0 g) of a compound represented by Formula (VIII).
  • The residual (500.0 g) of Formula (VIII) was dissolved in methanol and isopropyl alcohol (250.0 g and 1000.0 g). (+)-O,O'-di-pivaloyl-D-tartaric acid (D-DPTA) (636.0 g, 2.0 mole) was added into the solution containing the compound of Formula (VIII). After adding, the reaction mixture was heated to about 60°C for one hour and then cooled down slowly to room temperature (25°C). The wet cake (1,143.0 g) of a compound represented by Formula (IX) was obtained after filtration, and the dry cake (946.0 g) was obtained after drying. The obtained compound represented by Formula (IX) had purity of 99.8%, measured by ultra-performance liquid chromatography, and yield of 85% (from propiophenone).
  • The compound represented by Formula (IX) (557.0 g, 1.0 mole) was mixed and stirred with heptane (795 g), water (1,295 g) and sodium hydroxide (80.0 g, 2.0 mole) at ambient temperature under inert atmosphere until all solid dissolved. The heptane layer was separated from the aqua layer, and washed with water (100.0 g). The S-form of the compound of Formula (VIII) was generated in the heptane layer, and then mixed with isopropyl alcohol (200.0 g). 5% Palladium on charcoal (10 g) was added to the reaction mixture and kept under 0.4 to 0.5 MPa hydrogen pressure for 4 hours at 30°C to 35°C, and for 4 hours at 60°C to 65°C. The completion of reaction was checked by liquid chromatography. Palladium on charcoal was filtered and washed with isopropyl alcohol (200.0 g). The reaction filtrate was then concentrated under reduced pressure at 50°C to 80°C. The residual (138.5 g) of L-norephedrine was obtained with purity of 98.5% measured by ultra-performance liquid chromatography. The ratio of hydrogenated product ((1R,2S)-2-amino-1-phenyl-1-propanol:(1S,2S)-2-amino-1-phenyl-1-propanol) was 90:10. Yield of L-norephedrine from the compound represented by Formula (IX) was 91%.
  • Example 7: Preparation of L-norephedrine from phenylpropanolamine
  • Figure imgb0023
  • Phenylpropanolamine (PPA) (302.0 g, 2.0 mole) was mixed with isopropyl alcohol (IPA) (698.0 g) at 50°C to form a clear solution under inert atmosphere. (+)-Dibenzoyl-D-tartaric acid (D-DBTA) (178.6 g, 0.48 mole) was dissolved in IPA (774.1 g) and slowly charged into PPA/IPA solution over three hours at 25°C to 50°C. The solid was formed during charging. After charging, the reaction mixture was subjected to stirring at about 25°C for two hours. After filtration, the crude wet cake (346.5 g) of the compound represented by Formula (X) was obtained with enantiomeric excess (e.e.) purity of about 95.7%, measured by chiral liquid chromatography.
  • For recrystallization, the crude wet cake of the compound represented by Formula (X) (346.5) was dissolved in methanol (1,200 g) and heated to reflux (about 66°C) to distill out methanol (609.0 g) under inert atmosphere. The mixture solution was cooled down slowly to about 1°C to 2°C and hold for another two hours. The wet cake of the compound represented by Formula (X) was obtained after filtration, and the dry cake (258.5 g) of the compound represented by Formula (X) was obtained after drying (e.e. purity of about 99.99%, measured by chiral liquid chromatography).
  • The compound represented by Formula (X) (132.0 g, 0.4 mole) was mixed and stirred with cyclopentyl methyl ether (CPME) (250.0 g) and 2 M HCl(aq) (400.0 g) at 75°C under inert atmosphere until all solid dissolved. The CPME layer was separated from the aqua layer, and washed with 1 M HCl(aq) (100.0 g). L-norephedrine of Formula (II) was generated in the collected HCl(aq) layer, and then neutralized with 45% NaOH (82.5 g, 0.93 mole) and extracted with dichloromethane four times (213.0 g, 103.0 g, 100.0 g and 100.0 g, respectively). The collected dichloromethane layer was concentrated under reduced pressure to obtain the residual (66.0 g) of L-norephedrine, in which the yield was 38.0%, from phenylpropanolamine.
  • While some of the embodiments of the present disclosure have been described in detail above, it is, however, possible for those of ordinary skill in the art to make various modifications and changes to the embodiments shown without substantially departing from the teaching and advantages of the present disclosure. Such modifications and changes are encompassed in the scope of the present disclosure as set forth in the appended claims.

Claims (20)

  1. A compound represented by Formula (VI) below or a salt thereof for preparing lisdexamhetamine or a salt thereof:
    Figure imgb0024
    wherein R1 is hydrogen, an aliphatic group, C1-C6 alkoxyl, a C1-C6 alkylamino group, or an aromatic group, and PG is an amine protecting group.
  2. The compound of claim 1, wherein R1 is methyl and PG is benzyloxycarbonyl.
  3. A method for preparing lisdexamphetamine or a salt thereof, comprising reducing an O-acylation group of an intermediate compound represented by Formula (VI) below or a salt thereof through a catalytic hydrogenation to obtain the lisdexamphetamine or the salt thereof:
    Figure imgb0025
    wherein R1 is hydrogen, an aliphatic group, C1-C6 alkoxyl, a C1-C6 alkylamino group, or an aromatic group, and PG is an amine protecting group.
  4. The method of claim 3, wherein R1 is methyl and PG is benzyloxycarbonyl.
  5. The method of claim 3, wherein the catalytic hydrogenation is carried out by contacting the intermediate compound represented by Formula (VI) with a catalyst selected from the group consisting of palladium on charcoal, nickel, palladium, platinum, ruthenium, rhodium, and any combination thereof.
  6. The method of claim 3, wherein the reducing is carried out in the presence of a first solvent at a first temperature in a range of from 25°C to 80°C, and wherein the first solvent is selected from the group consisting of acetic acid, methanol, ethanol, isopropyl alcohol, water, dichloromethane, N-methylpyrrolidone, tetrahydrofuran, ethyl acetate, dimethylformamide, acetonitrile, dimethyl sulfoxide, toluene, hexane, ether, and any combination thereof.
  7. The method of claim 6, wherein a weight ratio of the intermediate compound represented by Formula (VI) to the first solvent is from 1:2 to 1:20.
  8. The method of claim 3, further comprising reacting a compound represented by Formula (V) below or a salt thereof with an acylation reagent to obtain the intermediate compound represented by Formula (VI):
    Figure imgb0026
    wherein PG is as defined in claim 3.
  9. The method of claim 8, wherein the acylation reagent is selected from the group consisting of formic acid, methyl formate, ethyl formate, acetic acid, acetic anhydride, ethyl acetate, acetyl chloride, trifluoroacetic acid, trifluoroacetic anhydride, benzoic acid, benzoic anhydride, benzoic methyl ester, benzoic chloride, acrylic acid, acryloyl chloride, di-tert-butyl dicarbonate, aryl chloroformate, and any combination thereof.
  10. The method of claim 9, wherein the acylation reagent is acetic anhydride having an equivalent of from 1.0 to 3.0.
  11. The method of claim 8, wherein the reacting is carried out in the presence of a first base at a second temperature in a range of from 25°C to 100°C, and wherein the first base is selected from the group consisting of imidazole, guanidine, N-methylmorpholine, diisopropylethyl amine (DIPEA), triethylamine (TEA), tri-n-propylamine, pyridine, 2,6-lutidine, piperidine, pyrrole, pyrrolidine, 1,8-diazabicyclo(5.4.0)undec-7-ene (DBU), and any combination thereof.
  12. The method of claim 11, wherein the first base is pyridine having an equivalent of from 1.0 to 3.0.
  13. The method of claim 8, further comprising coupling L-norephedrine represented by Formula (II) below or a salt thereof:
    Figure imgb0027
    and
    a diamino protected L-lysine represented by Formula (IV) below or a salt thereof:
    Figure imgb0028
    to obtain the compound represented by Formula (V), wherein PG is as defined in claim 3.
  14. The method of claim 13, wherein the coupling is carried out by reacting the L-norephedrine represented by Formula (II) or the salt thereof and the diamino protected L-lysine represented by Formula (IV) or the salt thereof with an acid activating reagent.
  15. The method of claim 14, wherein the acid activating reagent is selected from the group consisting of dicyclohexyl carbodiimide (DCC), 1-ethyl-3-(dimethylamino)carbodiimide hydrochloride (EDC-HCl), carbonyldiimidazole (CDI), benzotriazol-1-yloxytri(pyrrolidino)phosphonium hexafluorophosphate (PyBOP), (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate (HATU), O-(1H-benzotriazol-1-yl)-N,N,N,N-tetramethyluronium hexafluorophosphate (HBTU), N-hydroxybenzotriazole, N-hydroxysuccinimide (HOSu), methyl chloroformate, ethyl chloroformate, isopropyl chloroformate, isobutyl chloroformate, benzyl chloroformate, phenyl chloroformate, aryloxy chloroformate, and any combination thereof.
  16. The method of claim 15, wherein the acid activating reagent is isobutyl chloroformate having an equivalent of from 0.95 to 1.05.
  17. The method of claim 14, wherein the coupling is carried out in the presence of a second base and a second solvent at a third temperature in a range of from -20°C to 40°C.
  18. The method of claim 17, wherein the second solvent is selected from the group consisting of dichloromethane, N-methylpyrrolidone, tetrahydrofuran, ethyl acetate, acetone, dimethylformamide, acetonitrile, dimethyl sulfoxide, and any combination thereof, and the second base is selected from the group consisting of N-methylmorpholine, diisopropylethyl amine (DIPEA), triethylamine (TEA), tri-n-propylamine, pyridine, 1,8-diazabicyclo(5.4.0)undec-7-ene (DBU), and any combination thereof.
  19. The method of claim 18, wherein the second base is N-methylmorpholine having an equivalent of from 1.0 to 2.4.
  20. The method of claim 3, further comprising contacting the lisdexamphetamine with methanesulfonic acid to obtain lisdexamfetamine dimesylate.
EP21187139.7A 2021-07-22 2021-07-22 Compound and method for preparation of lisdexamfetamine Active EP4122914B1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP21187139.7A EP4122914B1 (en) 2021-07-22 2021-07-22 Compound and method for preparation of lisdexamfetamine
EP23156189.5A EP4215517A1 (en) 2021-07-22 2021-07-22 Compound and method for preparation of lisdexamfetamine

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP21187139.7A EP4122914B1 (en) 2021-07-22 2021-07-22 Compound and method for preparation of lisdexamfetamine

Related Child Applications (2)

Application Number Title Priority Date Filing Date
EP23156189.5A Division-Into EP4215517A1 (en) 2021-07-22 2021-07-22 Compound and method for preparation of lisdexamfetamine
EP23156189.5A Division EP4215517A1 (en) 2021-07-22 2021-07-22 Compound and method for preparation of lisdexamfetamine

Publications (2)

Publication Number Publication Date
EP4122914A1 true EP4122914A1 (en) 2023-01-25
EP4122914B1 EP4122914B1 (en) 2023-08-16

Family

ID=77021237

Family Applications (2)

Application Number Title Priority Date Filing Date
EP21187139.7A Active EP4122914B1 (en) 2021-07-22 2021-07-22 Compound and method for preparation of lisdexamfetamine
EP23156189.5A Pending EP4215517A1 (en) 2021-07-22 2021-07-22 Compound and method for preparation of lisdexamfetamine

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP23156189.5A Pending EP4215517A1 (en) 2021-07-22 2021-07-22 Compound and method for preparation of lisdexamfetamine

Country Status (1)

Country Link
EP (2) EP4122914B1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010042120A1 (en) 2008-10-09 2010-04-15 Archimica, Inc. Process for the synthesis of amphetamine derivatives
WO2010148305A1 (en) 2009-06-19 2010-12-23 Cambrex Charles City, Inc. Methods and compositions for preparation of amphetamine conjugates and salts thereof
US20120157706A1 (en) 2010-12-20 2012-06-21 Bauer Michael J Methods and Compositions for Preparing Lisdexamfetamine and Salts Thereof
WO2013011526A1 (en) 2011-07-20 2013-01-24 Ind-Swift Laboratories Limited Process for preparation of lisdexamphetamine and salts thereof
US20160376618A1 (en) 2015-06-29 2016-12-29 Noramco, Inc. Process for the preparation of lisdexamfetamine and related derivatives
WO2017098533A2 (en) 2015-12-11 2017-06-15 Sun Pharmaceutical Industries Limited Process for preparation of lisdexamphetamine

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010042120A1 (en) 2008-10-09 2010-04-15 Archimica, Inc. Process for the synthesis of amphetamine derivatives
WO2010148305A1 (en) 2009-06-19 2010-12-23 Cambrex Charles City, Inc. Methods and compositions for preparation of amphetamine conjugates and salts thereof
US20120157706A1 (en) 2010-12-20 2012-06-21 Bauer Michael J Methods and Compositions for Preparing Lisdexamfetamine and Salts Thereof
WO2013011526A1 (en) 2011-07-20 2013-01-24 Ind-Swift Laboratories Limited Process for preparation of lisdexamphetamine and salts thereof
US20160376618A1 (en) 2015-06-29 2016-12-29 Noramco, Inc. Process for the preparation of lisdexamfetamine and related derivatives
WO2017098533A2 (en) 2015-12-11 2017-06-15 Sun Pharmaceutical Industries Limited Process for preparation of lisdexamphetamine

Also Published As

Publication number Publication date
EP4122914B1 (en) 2023-08-16
EP4215517A1 (en) 2023-07-26

Similar Documents

Publication Publication Date Title
CA2176012C (en) Novel phenylalkylaminoalcohol carbamates and process for preparing the same
US20090312548A1 (en) Modified pictet-spengler reaction and products prepared therefrom
EP0581167B1 (en) Novel arylglycinamide derivatives and preparative processes therefor and their use for treatment of dysuria
KR101456839B1 (en) Compositions and methods of making a photoactive agent
EP0340064B1 (en) Benzodiazepines, process and intermediates for their preparation and their use in therapy
OTSUKA et al. Synthetic studies on an antitumor antibiotic, Bleomycin. XII. Preparation of an l-2, 3-diaminopropionic acid derivative as a synthetic intermediate
RU2715233C2 (en) Method of producing nitrogen mustard derivatives
JP2000502364A (en) O-carbamoyl-phenylalaninol compound, pharmaceutically useful salt thereof and method for producing the same
EP2736909A1 (en) Process for the preparation of sitagliptin and its pharmaceutically acceptable salts
EP4122914B1 (en) Compound and method for preparation of lisdexamfetamine
EP0741697B1 (en) Amino acid derivatives and their use as phospholipase a2 inhibitors
US11608312B1 (en) Compound and method for preparation of lisdexamfetamine
KR101343443B1 (en) Colchicine derivatives or phamarceutically acceptable salts thereof, process for preparation thereof and pharmaceutical composition containing the same
AU761721B2 (en) Method for producing enantiomer-free N-methyl-N- ((1S)-1-phenyl- 2-((3S)- 3-hydroxypyrrolidine- 1-yl)ethyl)- 2,2-diphenyl acetamide
EP2383255A1 (en) New compounds, synthesis and use thereof in the treatment of pain
US20030083344A1 (en) Tryptase inhibitors
Szakonyi et al. Mild and efficient ring opening of monoterpene-fused β-lactam enantiomers. Synthesis of novel β-amino acid derivatives
EP2058300B1 (en) Succinic acid diester derivative, process for production thereof, and use of the derivative in the production of pharmaceutical preparation
Rutar et al. Enantiomers of 2‐methyl‐and 2, 4‐dimethyl‐3‐oxo‐3, 4‐dihydro‐2H‐1, 4‐benzoxazine‐2‐carboxylic acid: Preparation by resolution, determination of absolute configuration, and Curtius rearrangement
EP2530072A1 (en) New compounds, synthesis and use thereof in the treatment of pain
FR2477142A1 (en) NOVEL DERIVATIVES OF NITROSOUREE, PROCESS FOR PREPARING THEM AND THEIR THERAPEUTIC APPLICATION
FR2497800A1 (en) NEW NITROSOUREE DERIVATIVES
KR100711938B1 (en) Method for preparing stereoselective 4-hydroxy-2-pyrrolidinone
Matulevičiūtė et al. Facile synthesis of new N-(aminocycloalkylene) amino acid compounds using chiral triflate esters with N-Boc-aminopyrrolidines and N-Boc-aminopiperidines
Pan et al. Diastereospecific synthesis of trans-2, 3-diaryl-1-aminocyclopropanecarboxylic acids

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN PUBLISHED

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20230209

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

RIC1 Information provided on ipc code assigned before grant

Ipc: C07C 271/22 20060101ALI20230419BHEP

Ipc: C07C 269/06 20060101ALI20230419BHEP

Ipc: C07C 237/06 20060101ALI20230419BHEP

Ipc: C07C 231/12 20060101AFI20230419BHEP

RAP3 Party data changed (applicant data changed or rights of an application transferred)

Owner name: SCI PHARMTECH INC

INTG Intention to grant announced

Effective date: 20230517

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602021004291

Country of ref document: DE

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG9D

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20231109

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20230816

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1599940

Country of ref document: AT

Kind code of ref document: T

Effective date: 20230816

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20231117

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20231216

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20231218

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20231116

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20231216

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20231117

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230816